Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Nucl Med Mol Imaging ; 39(12): 1886-97, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872310

ABSTRACT

PURPOSE: Targeted therapy with α-particle emitting radionuclides is a promising new option in cancer therapy. Stable conjugates of the vascular tumour-homing peptide F3 with the α-emitter (213)Bi specifically target tumour cells. The aim of our study was to determine efficacy of combined (213)Bi-diethylenetriaminepentaacetic acid (DTPA)-F3 and paclitaxel treatment compared to treatment with either (213)Bi-DTPA-F3 or paclitaxel both in vitro and in vivo. METHODS: Cytotoxicity of treatment with (213)Bi-DTPA-F3 and paclitaxel, alone or in combination, was assayed towards OVCAR-3 cells using the alamarBlue assay, the clonogenic assay and flow cytometric analyses of the mode of cell death and cell cycle arrest. Therapeutic efficacy of the different treatment options was assayed after repeated treatment of mice bearing intraperitoneal OVCAR-3 xenograft tumours. Therapy monitoring was performed by bioluminescence imaging and histopathologic analysis. RESULTS: Treatment of OVCAR-3 cells in vitro with combined (213)Bi-DTPA-F3 and paclitaxel resulted in enhanced cytotoxicity, induction of apoptosis and G2/M phase arrest compared to treatment with either (213)Bi-DTPA-F3 or paclitaxel. Accordingly, i.p. xenograft OVCAR-3 tumours showed the best response following repeated (six times) combined therapy with (213)Bi-DTPA-F3 (1.85 MBq) and paclitaxel (120 µg) as demonstrated by bioluminescence imaging and histopathologic investigation of tumour spread on the mesentery of the small and large intestine. Moreover, mean survival of xenograft mice that received combined therapy with (213)Bi-DTPA-F3 and paclitaxel was significantly superior to mice treated with either (213)Bi-DTPA-F3 or paclitaxel alone. CONCLUSION: Combined treatment with (213)Bi-DTPA-F3 and paclitaxel significantly increased mean survival of mice with peritoneal carcinomatosis of ovarian origin, thus favouring future therapeutic application.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma/therapy , Chemoradiotherapy , Organometallic Compounds/therapeutic use , Paclitaxel/therapeutic use , Peritoneal Neoplasms/therapy , Radiopharmaceuticals/therapeutic use , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints/drug effects , HEK293 Cells , HMGN2 Protein/chemistry , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Organometallic Compounds/pharmacology , Paclitaxel/pharmacology , Protein Structure, Tertiary , Radiopharmaceuticals/pharmacology , Treatment Outcome
2.
Cancer Res ; 72(19): 5014-24, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22875026

ABSTRACT

The prognosis of relapsed or refractory aggressive lymphoma is poor. The huge variety of currently evolving targeted treatment approaches would benefit from tools for early prediction of response or resistance. We used various ALK-positive anaplastic large cell lymphoma (ALCL) cell lines to evaluate two inhibitors, the HSP90 inhibitor NVP-AUY922, and the mTOR inhibitor everolimus, both of which have shown to interfere with ALK-dependent oncogenic signal transduction. Their therapeutic effect was determined in vitro by MTT assay, [(18)F]fluorodeoxyglucose (FDG)- and [(18)F]fluorothymidine (FLT)-uptake, and by biochemical analysis of ALK-induced signaling. Micro-FDG- and FLT-positron emission tomography (PET) imaging studies in immunodeficient mice bearing ALCL xenotransplants were carried out with the cell lines SUDHL-1 and Karpas299 to assess early treatment response to NVP-AUY922 or everolimus in vivo. SUDHL-1 cells showed sensitivity to both inhibitors in vitro. Importantly, we detected a significant reduction of FLT-uptake in SUDHL-1 bearing animals using both inhibitors compared with baseline as early as 5 days after initiation of targeted therapy. Immunostaining showed a decrease in Ki-67 and an increase in cleaved caspase-3 staining. In contrast, FDG-uptake did not significantly decrease at early time points. Karpas299 xenotransplants, which are resistant to NVP-AUY922 and sensitive to everolimus treatment, showed an increase of mean FLT-uptake on day 2 after administration of NVP-AUY299, but a significant reduction in FLT-uptake upon everolimus treatment. In conclusion, we show that FLT-PET but not FDG-PET is able to predict response to treatment with specific inhibitors very early in the course of treatment and thus enables early prediction of treatment efficacy.


Subject(s)
Enzyme Inhibitors/pharmacology , Lymphoma, Large-Cell, Anaplastic/diagnostic imaging , Lymphoma, Large-Cell, Anaplastic/drug therapy , Positron-Emission Tomography/methods , Xenograft Model Antitumor Assays , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Dideoxynucleosides , Everolimus , Female , Flow Cytometry , Fluorine Radioisotopes , Fluorodeoxyglucose F18 , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Isoxazoles/pharmacology , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , Mice, SCID , Protein-Tyrosine Kinases/metabolism , Resorcinols/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Tumor Burden/drug effects
3.
Biochem J ; 445(1): 135-44, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22509934

ABSTRACT

ADAM17 (A disintegrin and metalloproteinase 17) is a membrane-bound protease that cleaves various cell surface proteins, including cytokines and cytokine receptors. Recently it was shown that ADAM17 is highly expressed on the surface of many cancer cells, whereas normal cells express low levels of ADAM17, implying that ADAM17 is a potential immunotherapeutic target. We have generated a monoclonal antibody against human ADAM17, which recognized the membrane proximal cysteine-rich extension of the ADAM17 protein. Unlike normal cells, tumour cell lines, such as a prostate cancer cell line, pancreatic cancer cell lines, a breast cancer cell line and a non-small lung cancer cell line, expressed ADAM17 on the cell surface. Using the sequence of the antibody we generated an ADAM17-specific scFv (single-chain variable fragment) and fused this to a CD3-specific scFv to generate a bispecific T-cell engager antibody [A300E-BiTE (bispecific T-cell engager antibody)]. Specificity was demonstrated on cells in which ADAM17 was knocked down with a specific shRNA (short hairpin RNA). A300E-BiTE recognized ADAM17 and CD3 on the cell surface of tumour cells and T-cells respectively. In the presence of primary human peripheral blood mononuclear cells or human T-cells the addition of A300E-BiTE led to ADAM17-specific killing of prostate tumour cells indicating a novel strategy for the treatment of cancer.


Subject(s)
ADAM Proteins/metabolism , Antibodies, Bispecific/pharmacology , CD3 Complex/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Single-Chain Antibodies/immunology , T-Lymphocytes/metabolism , ADAM Proteins/immunology , ADAM17 Protein , Antibodies, Bispecific/immunology , Apoptosis , Blotting, Western , CD3 Complex/immunology , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoprecipitation , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Prostatic Neoplasms/immunology , RNA, Messenger/genetics , T-Lymphocytes/immunology , Tumor Cells, Cultured
4.
Oncotarget ; 2(8): 599-609, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21865609

ABSTRACT

Squamous cell cancer of the head and neck (SCCHN) is the sixth leading cause for cancer deaths worldwide. Despite extense knowledge of risk factors and pathogenesis about 50 percent of all patients and essentially every patient with metastatic SCCHN eventually die from this disease. We analyzed the clinical data and performed immunohistochemistry for Epidermal growth factor receptor (EGFR) and Aurora kinase A (Aurora-A) expression in 180 SCCHN patients. Patients characterized by elevated EGFR and elevated Aurora-A protein expression in tumor tissue represent a risk group with poor disease-free and overall survival (EGFR(low)Aurora-A(low) versus EGFR(high)Aurora-A(high), p = 0.024). Treating SCCHN cell lines with a pan-Aurora kinase inhibitor resulted in defective cytokinesis, polyploidy and apoptosis, which was effective irrespective of the EGFR status. Combined Aurora kinase and EGFR targeting using a monoclonal anti-EGFR antibody was more effective compared to single EGFR and Aurora kinase inhibition. Comparing pan-Aurora kinase and Aurora-A targeting hints towards a strong and clinically relevant biological effect mediated via Aurora kinase B. Taken together, our findings characterize a new poor risk group in SCCHN patients defined by elevated EGFR and Aurora-A protein expression. Our results demonstrate that combined targeting of EGFR and Aurora kinases represents a therapeutic means to activate cell cycle checkpoints and apoptosis in SCCHN.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/enzymology , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/enzymology , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal, Humanized , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Cell Line, Tumor , Cetuximab , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/surgery , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Protein Serine-Threonine Kinases/metabolism , Survival Rate , Time Factors , Up-Regulation
5.
Cancer Res ; 70(13): 5368-78, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20551051

ABSTRACT

Epidermal growth factor receptor (EGFR) overexpression and activation are hallmarks of non-small cell lung carcinoma (NSCLC). Although EGFR-targeted therapies are used, the prognosis of NSCLC remains poor. ADAM17 induces activation of the EGFR through ligand cleavage. However, we show that inhibition or knockdown of ADAM17 markedly reduces tumorigenesis and survival to a large part independently from EGFR ligand shedding in NSCLC cells. These findings strongly indicate additional oncogenic mechanisms regulated by ADAM17. We identified Notch1 signaling as an ADAM17-controlled pathway and a critical regulator of anchorage-independent growth by using both Notch1 shRNA and ectopic expression of the active intracellular Notch1 fragment. Strikingly, Notch1 knockdown led to a strong reduction of EGFR expression in all analyzed cell lines. Proliferation, survival, and colony formation of Notch1-deficient cells were insensitive to EGF stimulation. Moreover, targeting Notch1 or ADAM17 resulted in substantial cell death, whereas EGFR inhibition predominantly induced cell cycle arrest. Immunohistochemical analysis of primary human tissue revealed a significant correlation between ADAM17, Notch1 signaling, and high EGFR expression levels. In conclusion, this article describes a novel molecular circuitry in NSCLC, incorporating ADAM17 as a regulator of EGFR expression through the activation of Notch1. Due to their central role in tumorigenesis and survival of NSCLC cells, both ADAM17 and Notch1 constitute promising targets for the treatment of NSCLC.


Subject(s)
ADAM Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/biosynthesis , Lung Neoplasms/metabolism , Receptor, Notch1/metabolism , ADAM Proteins/biosynthesis , ADAM17 Protein , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epidermal Growth Factor/pharmacology , Homeodomain Proteins/biosynthesis , Humans , Lung Neoplasms/pathology , Mice , Receptor, Notch1/biosynthesis , Signal Transduction , Transcription Factor HES-1 , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...